487
Views
26
CrossRef citations to date
0
Altmetric
Review

Acetylcholinesterase inhibitors for the treatment of Alzheimer’s disease – a patent review (2016–present)

, , , ORCID Icon, ORCID Icon, , ORCID Icon & ORCID Icon show all
Pages 399-420 | Received 21 Sep 2020, Accepted 07 Jan 2021, Published online: 14 Jan 2021

References

  • [cited 2007 Sep 30]. Available from: https://www.who.int/news-room/fact-sheets/detail/dementia.
  • Patterson C. World Alzheimer report 2018. In: the state of the art of dementia research: new frontiers. Alzheimer’s Disease International. London, UK: Alzheimer’s Disease International (ADI); 2018. p. 32–36.
  • Nichols E, Szoeke CEI, Vollset SE, et al., Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 18(1): 88–106. 2019. .
  • Fan J, Tao W, Li X, et al. The contribution of genetic factors to cognitive impairment and dementia: apolipoprotein e gene, gene interactions, and polygenic risk. Int J Mol Sci. 2019;20(5):1177.
  • Yuksel M, Tacal O. Trafficking and proteolytic processing of amyloid precursor protein and secretases in Alzheimer’s disease development: an up-to-date review. Eur J Pharmacol. 2019;856:172415.
  • Chen Y, Fu AK, Ip NY. Synaptic dysfunction in Alzheimer’s disease: mechanisms and therapeutic strategies. Pharmacol Ther. 2019;195:186–198.
  • Kumar A, Singh A. A review on Alzheimer’s disease pathophysiology and its management: an update. Pharmacol Rep. 2015;67(2):195–203.
  • Mroczko B, Groblewska M, The L-ZA. Role of Protein Misfolding and Tau Oligomers (TauOs) in Alzheimer′ s Disease (AD). Int J Mol Sci. 2019;20(19):4661.
  • Contestabile A. The history of the cholinergic hypothesis. Behav Brain Res. 2011;221(2):334–340.
  • Brus B, Kosak U, Turk S, et al. Discovery, biological evaluation, and crystal structure of a novel nanomolar selective butyrylcholinesterase inhibitor. J Med Chem. 2014;57(19):8167–8179. .
  • Rosenberry TL, Brazzolotto X, Macdonald IR, et al., Comparison of the binding of reversible inhibitors to human butyrylcholinesterase and acetylcholinesterase: A crystallographic, kinetic and calorimetric study. Molecules. 22(12): 2098. 2017. .
  • Johnson G, Moore S. The peripheral anionic site of acetylcholinesterase: structure, functions and potential role in rational drug design. Current Pharmaceutical Design. 2006;12(2):217–225.
  • Cheung J, Rudolph MJ, Burshteyn F, et al. Structures of human acetylcholinesterase in complex with pharmacologically important ligands. J Med Chem. 2012;55(22):10282–10286. .
  • Inestrosa NC, Dinamarca MC, Alvarez A. Amyloid-cholinesterase interactions. Febs J. 2008;275(4):625–632.
  • Bartolini M, Bertucci C, Cavrini V, et al. β-Amyloid aggregation induced by human acetylcholinesterase: inhibition studies. Biochem Pharmacol. 2003;65(3):407–416.
  • Silman I, Sussman JL. Acetylcholinesterase: ‘classical’ and ‘non-classical’ functions and pharmacology. Curr Opin Pharmacol. 2005;5(3):293–302.
  • Alzheimer's Association Report. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement. 2020; 16(3): 391–460.
  • Pandolfi F, De Vita D, Bortolami M, et al. New pyridine derivatives as inhibitors of acetylcholinesterase and amyloid aggregation. Eur J Med Chem. 2017;141:197–210.
  • Bortolami M, Pandolfi F, De Vita D, et al. New deferiprone derivatives as multi-functional cholinesterase inhibitors: design, synthesis and in vitro evaluation. Eur J Med Chem. 2020;198:112350.
  • Gong C-X, Liu F, Iqbal K. Multifactorial hypothesis and multi-targets for Alzheimer’s disease. Journal of Alzheimer’s Disease. 2018;64(s1):S107–S17.
  • Albertini C, Salerno A, de Sena Murteira Pinheiro P. From combinations to multitarget-directed ligands: A continuum in Alzheimer’s disease polypharmacology. Med Res Rev. 2020. 10.1002/med.21699
  • Kabir M, Uddin M, Mamun AA, et al. Combination drug therapy for the management of Alzheimer’s disease. Int J Mol Sci. 2020;21(9):3272. .
  • Wang H, Zhang H. Reconsideration of anticholinesterase therapeutic strategies against Alzheimer’s disease. ACS Chem Neurosci. 2019;10(2):852–862. .
  • Panek D, Wichur T, Godyń J, et al. Advances toward multifunctional cholinesterase and β-amyloid aggregation inhibitors. Future Med Chem. 2017;9(15):1835–1854.
  • Rosini M, Simoni E, Caporaso R, et al. Multitarget strategies in Alzheimer’s disease: benefits and challenges on the road to therapeutics. Future Med Chem. 2016;8(6):697–711.
  • de Los Ríos C. Cholinesterase inhibitors: a patent review (2007 – 2011). Expert Opin Ther Pat. 2012;22(8):853–869.
  • Tumiatti V, Bolognesi ML, Minarini A, et al. Progress in acetylcholinesterase inhibitors for Alzheimer’s disease: an update. Expert Opin Ther Pat. 2008;18(4):387–401. .
  • Pohanka M. Acetylcholinesterase inhibitors: a patent review (2008 – present). Expert Opin Ther Pat. 2012;22(8):871–886.
  • McHardy SF, Wang H-YL, McCowen SV, et al. Recent advances in acetylcholinesterase Inhibitors and Reactivators: an update on the patent literature (2012-2015).. Expert Opin Ther Pat. 2017;27(4):455–476.
  • Zálešák F, Bon DJ-YD, Pospíšil PJ. Lignans and Neolignans: plant secondary metabolites as a reservoir of biologically active substances. Pharmacol Res. 2019;146:104284.
  • Qi Y, Dou D-Q, Jiang H, et al. Arctigenin attenuates learning and memory deficits through pi3k/akt/gsk-3β pathway reducing tau hyperphosphorylation in aβ-induced ad mice.. Planta Med. 2017;83(1–02):51–56. .
  • Zhu Z, Yan J, Jiang W, et al. Arctigenin effectively ameliorates memory impairment in alzheimer’s disease model mice targeting both -amyloid production and clearance. J Neurosci. 2013;33(32):13138–13149. .
  • Gao Q, Yang M, Zuo Z. Overview of the anti-inflammatory effects, pharmacokinetic properties and clinical efficacies of arctigenin and arctiin from Arctium lappa L. Acta Pharmacol Sin. 2018;39(5):787–801.
  • Jilin agricultural university. Acetylcholinesterase inhibitor lignanolide compound and preparation method and application thereof. CN105541764A (2016).
  • Nano egg KK. acetylcholinesterase (AChE) inhibitors. JP2018062478A (2018).
  • Miura Y, Gotoh E, Nara F, et al. Hydrolysis of sphingosylphosphocholine by neutral sphingomyelinases. FEBS Lett. 2004;557(1–3):288–292.
  • Yangzhou polytechnic institut.Preparation method of isocoumarin long-chain carboxylic ester compound and application thereof. CN108586418A (2018).
  • Rook Y, Schmidtke K-U, Gaube F, et al. Bivalent β-carbolines as potential multitarget anti-Alzheimer agents. J Med Chem. 2010;53(9):3611–3617.
  • Horton W, Sood A, Peerannawar S, et al. Synthesis and application of β-carbolines as novel multi-functional anti-Alzheimer’s disease agents. Bioorg Med Chem Lett. 2017;27(2):232–236.
  • Zhao Y, Ye F, Xu J, et al. Design, synthesis and evaluation of novel bivalent β-carboline derivatives as multifunctional agents for the treatment of Alzheimer’s disease. Bioorg Med Chem. 2018;26(13):3812–3824.
  • Univ Northwest A&F. Carboline compounds, and synthetic method and anti-acetylcholinesterase activity thereof. CN106588917A (2017).
  • Dai J, Dan W, Schneider U, et al. β-Carboline alkaloid monomers and dimers: occurrence, structural diversity, and biological activities. Eur J Med Chem. 2018;157:622–656.
  • Ercetin Tugba.1 -(dimethylamino)ethyl-substituted 6H-benzo[c]chromen-6-ones against senile dementia. US2016002194A1 (2016).
  • Landete J. Ellagitannins, ellagic acid and their derived metabolites: a review about source, metabolism, functions and health. Food Res Int. 2011;44(5):1150–1160.
  • Prasher P, Sharma M. Medicinal chemistry of acridine and its analogues. MedChemComm. 2018;9(10):1589–1618.
  • Gensicka-Kowalewska M, Cholewiński G, Dzierzbicka K. Recent developments in the synthesis and biological activity of acridine/acridone analogues. RSC Adv. 2017;7(26):15776–15804.
  • Guangxi university of chinese medicine. application of 2-methoxy-9-acridine(4-methoxybenzamido) thiourea to preparing of acetylcholin esterase. CN105412108A (2016).
  • Henan normal university.Acridone derivative containing amino sugar structure and preparation method and application of acridone derivative. CN110642899A (2020).
  • Huaihai institute of technology. Preparation method and application of ethyl 2-(4-hydroxyphenyl)thiazole-4-carboxylate derivatives. CN106749090A (2017).
  • Zhejiang hisun pharm co ltd. Benzodioxole derivative and preparation method and use thereof. ES2645643T3 (2017).
  • International healthcare innovation institute. Pyrazol compound with acetylcholinesterase inhibition activity as well as preparation method and application thereof. CN108218775A (2018)
  • International healthcare innovation institute. Indeno[1,2-c] pyrazol compound and preparation method and application thereof. CN107868052A (2018).
  • International healthcare innovation institute, wuyi university. Novel application of tetrahydro cyclopenta pyrrole compound and derivatives of tetrahydro cyclopenta pyrrole. CN108371660A (2018).
  • International healthcare innovation institute, wuyi university. Nitrogenous heterocyclic quaternary carbon spiro compound and preparation method and application thereof. CN108003159A (2018).
  • Jiangmen dajiankang international innovation institute, wuyi university. Novel application of dihydrocomosidine spiro-compounds. CN107951884A (2018).
  • Sameem B, Saeedi M, Mahdavi M, et al. A review on tacrine-based scaffolds as multi-target drugs (MTDLs) for Alzheimer’s disease. Eur J Med Chem. 2017;128:332–345.
  • Shandong university. Aldoxime-containing tacrine derivative selective butyrylcholine esterase inhibitors, and preparation method and application thereof. CN111592530A (2020).
  • Kannur university. Tacrine derivatives targeting NMDA receptor, acetylcholine esterase, butyryl choline and beta secretase activity. WO2019207604A1 (2019).
  • Hershkowitz N, Rogawski MA. Tetrahydroaminoacridine block of N-methyl-D-aspartate-activated cation channels in cultured hippocampal neurons. Mol Pharmacol. 1991;39(5):592–598.
  • Armada-Moreira A, Gomes JI, Pina CC, et al. Going the extra (synaptic) mile: excitotoxicity as the road toward neurodegenerative diseases. Front Cell Neurosci. 2020;14:90.
  • Wang R, Reddy PH. Role of glutamate and NMDA receptors in Alzheimer’s disease. J Alzheimers Dis. 2017;57(4):1041–1048.
  • Viayna E, Sola I, Bartolini M, et al. Synthesis and multitarget biological profiling of a novel family of rhein derivatives as disease-modifying anti-Alzheimer agents. J Med Chem. 2014;57(6):2549–2567.
  • Serrano FG, Tapia-Rojas C, J F CP, et al. Rhein-huprine derivatives reduce cognitive impairment, synaptic failure and amyloid pathology in AβPPswe/PS-1 mice of different ages. Curr Alzheimer Res. 2016;13(9):1017–1029.
  • Pontificia Universidad Católica De Chile, Univ Barcelona. Beta-amyloid-directed multitarget compounds for the treatment of Alzheimer’s disease. . US9238626B2 (2016).
  • Univ Barcelona. Multitarget compounds for the treatment of Alzheimer’s disease. WO2020193448A1 (2020).
  • Morisseau C, Hammock BD. Impact of soluble epoxide hydrolase and epoxyeicosanoids on human health. Annu Rev Pharmacol Toxicol. 2013;53:37–58.
  • Griñán-Ferré C, Codony S, Pujol E, et al. Pharmacological inhibition of soluble epoxide hydrolase as a new therapy for Alzheimer’s disease. Neurotherapeutics. 2020;605055.
  • China pharmaceutical university. GSK-3beta/ChE double inhibitor as well as preparation method and application thereof. CN111004228A.(2020).
  • Sivaprakasam P, Han X, Civiello RL, et al. Discovery of new acylaminopyridines as GSK-3 inhibitors by a structure guided in-depth exploration of chemical space around a pyrrolopyridinone core. Bioorg Med Chem Lett. 2015;25(9):1856–1863.
  • China pharmaceutical university. Pyrimidinone compound or pharmaceutically acceptable salt, and preparation method and application thereof. CN111349085A.(2020).
  • Universidad autonoma de madrid. Use of the compounds derived from 3-alkyl-4-aryl-1,4,6,7,8,9-hexahydropyrazolo [4 ‘, 3ʹ: 5,6] pyran [2,3-b] quinolin-5- amine as dual inhibitors of gsk3b-ache and inducers of nrf2 for the treatment of neurodegenerative diseases. ES2587137A2 (2016).
  • Bahn G, Jo D-G. Therapeutic approaches to Alzheimer’s disease through modulation of NRF2. Neuromolecular Med. 2019;21(1):1–11.
  • Deepalakshmi K, Sankaran M. Pleurotus ostreatus: an oyster mushroom with nutritional and medicinal properties. J Biochem Tech 2014;5(2):718–726.
  • Tong H, Xia F, Feng K, et al. Structural characterization and in vitro antitumor activity of a novel polysaccharide isolated from the fruiting bodies of Pleurotus ostreatus. Bioresour Technol. 2009;100(4):1682–1686.
  • Facchini JM, Alves EP, Aguilera C, et al. Antitumor activity of pleurotus ostreatus polysaccharide fractions on ehrlich tumor and sarcoma 180. Int J Biol Macromol. 2014;68:72–77.
  • Zhang Y, Dai L, Kong X, et al. Characterization and in vitro antioxidant activities of polysaccharides from pleurotus ostreatus. Int J Biol Macromol. 2012;51(3):259–265.
  • Jilin institute of chemical technology. Application of pleutotus ostreatus polysaccharide. CN105902560A (2016).
  • Univ tianjin technology. Extraction method and medical application of guanidine alkaloids Buthus martensii Karsch alkaloid A and/or Buthus martensii Karsch alkaloid B in scorpions. CN110872240A (2020).
  • Prakash A, Dhaliwal GK, Kumar P, et al. Brain biometals and Alzheimer’s disease–boon or bane? Int J Neurosci. 2017;127(2):99–108.
  • Chen K-L, Gan L, Wu Z-H, et al. 4-Substituted sampangine derivatives: novel acetylcholinesterase and β-myloid aggregation inhibitors. Int J Biol Macromol. 2018;107:2725–2729.
  • Agarwal AK, Xu T, Jacob MR, et al. Role of heme in the antifungal activity of the azaoxoaporphine alkaloid sampangine. Eukaryot Cell. 2008;7(2):387–400.
  • Kluza J, Mazinghien R, Degardin K, et al. Induction of apoptosis by the plant alkaloid sampangine in human HL-60 leukemia cells is mediated by reactive oxygen species. Eur J Pharmacol. 2005;525(1–3):32–40.
  • Mahdi F, Morgan JB, Liu W, et al. Sampangine (a copyrine alkaloid) exerts biological activities through cellular redox cycling of its quinone and semiquinone intermediates. J Nat Prod. 2015;78(12):3018–3023.
  • Su C, Chen Y, Chen K, et al. Inhibitory potency of 4-substituted sampangine derivatives toward Cu2+ mediated aggregation of amyloid β-peptide, oxidative stress, and inflammation in Alzheimer’s disease. Neurochem Int. 2020;139:104794.
  • Peterson JR, Zjawiony JK, Liu S, et al. Copyrine alkaloids: synthesis, spectroscopic characterization, and antimycotic/antimycobacterial activity of A-and B-ring-functionalized sampangines. J Med Chem. 1992;35(22):4069–4077.
  • Claes P, Cappoen D, Mbala BM, et al. Synthesis and antimycobacterial activity of analogues of the bioactive natural products sampangine and cleistopholine. Eur J Med Chem. 2013;67:98–110.
  • University guangxi normal. 4-substituted Sampangine alkaloid derivative as well as synthesis method and application thereof. CN106905317A (2017).
  • Guangxi normal university. Application of 11-substituted oxoisoaporphine derivatives. CN106038564A (2016).
  • Castro-Castillo V, Rebolledo-Fuentes M, Theoduloz C, et al. Synthesis of lakshminine and antiproliferative testing of related oxoisoaporphines. J Nat Prod. 2010;73(11):1951–1953.
  • Zhang J, Chen L, Oxoisoaporphine Alkaloids SJ. Prospective anti‐alzheimer’s disease, anticancer, and antidepressant agents. ChemMedChem. 2018;13(13):1262–1274.
  • Di L, Kerns EH, Fan K, et al. High throughput artificial membrane permeability assay for blood–brain barrier. Eur J Med Chem. 2003;38(3):223–232.
  • Zuccarello E, Acquarone E, Calcagno E, et al. Development of novel phosphodiesterase 5 inhibitors for the therapy of Alzheimer’s disease. Biochem Pharmacol. 2020;176:113818.
  • Central south university. 2-Benzoyl-6-amino-4(3H)-quinazolinone derivatives and synthetic method and application thereof. CN109897008A.(2019).
  • Rodriguez JJ, Noristani HN, Verkhratsky A. The serotonergic system in ageing and Alzheimer’s disease. Prog Neurobiol. 2012;99(1):15–41.
  • Geldenhuys WJ. Van der Schyf CJ. Role of serotonin in Alzheimer’s disease. CNS Drugs. 2011;25(9):765–781.
  • Lanthier C, Dallemagne P, Lecoutey C, et al. Therapeutic modulators of the serotonin 5-HT4 receptor: a patent review (2014-present). Expert Opin Ther Pat. 2020;30(7):495–508.
  • Bockaert J, Claeysen S, Compan V, et al. 5-HT4 receptors, a place in the sun: act two. Curr Opin Pharmacol. 2011;11(1):87–93.
  • King MV, Marsden CA, Fone KC. A role for the 5-HT1A, 5-HT4 and 5-HT6 receptors in learning and memory. Trends Pharmacol Sci. 2008;29(9):482–492.
  • Universite de caen. acetylcholinesterase inhibitor compounds and 5ht4 serotonergic receptor agonists, with promnesia effect, Methods for the Preparation Thereof and Pharmaceutical Compositions Containing Same. US2016122300A1 (2016).
  • Lecoutey C, Hedou D, Freret T, et al. Design of donecopride, a dual serotonin subtype 4 receptor agonist/acetylcholinesterase inhibitor with potential interest for Alzheimer’s disease treatment. Proc Nat Acad Sci. 2014;111(36):E3825–E30. .
  • Rochais C, Lecoutey C, Gaven F, et al. Novel multitarget-directed ligands (mtdls) with acetylcholinesterase (ache) inhibitory and serotonergic subtype 4 receptor (5-ht 4 r) agonist activities as potential agents against alzheimer’s disease: the design of donecopride. J Med Chem. 2015;58(7):3172–3187. .
  • Rochais C, Lecoutey C, Hamidouche K, et al. Donecopride, a Swiss army knife with potential against Alzheimer’s disease. Br J Pharmacol. 2020;177(9):1988–2005. .
  • Univ china pharma. Donepezil-BHT heterozygotes and preparation method and application thereof in treatment of Alzheimer’s disease. CN109134350A (2019).
  • Cai Z. Monoamine oxidase inhibitors: promising therapeutic agents for Alzheimer’s disease (review). Mol Med Rep. 2014;9(5):1533–1541.
  • Gülcan HO, Orhan IE. The main targets involved in neuroprotection for the treatment of alzheimer’s disease and parkinson disease. Curr Pharm Des. 2020;26(4):509–516.
  • Mathew B, Parambi DGT, Mathew GE, et al., Emerging therapeutic potentials of dual-acting MAO and AChE inhibitors in Alzheimer’s and Parkinson’s diseases. Arch Pharm (Weinheim). 352(11): 1900177. 2019. .
  • Univ east china science & tech.Mono-propargylamine modified thiouracil compound and applications thereof. CN106946854A (2017).
  • Wang Y, Sun Y, Guo Y, et al. Dual functional cholinesterase and MAO inhibitors for the treatment of Alzheimer’s disease: synthesis, pharmacological analysis and molecular modeling of homoisoflavonoid derivatives.. J Enzyme Inhib Med Chem. 2016;31(3):389–397.
  • Joubert J, Foka GB, Repsold BP, et al. Synthesis and evaluation of 7-substituted coumarin derivatives as multimodal monoamine oxidase-B and cholinesterase inhibitors for the treatment of Alzheimer’s disease. Eur J Med Chem. 2017;125:853–864.
  • Xu Y, Zhang J, Wang H, et al. Rational design of novel selective dual-target inhibitors of acetylcholinesterase and monoamine oxidase B as potential anti-Alzheimer’s disease agents. ACS Chem Neurosci. 2019;10(1):482–496. .
  • Li Q, He S, Chen Y, et al. Donepezil-based multi-functional cholinesterase inhibitors for treatment of Alzheimer’s disease. Eur J Med Chem. 2018;158:463–477.
  • Suven life sciences ltd.Polycyclic amides as muscarinic M1 receptor positive allosteric modulatorsCA3078164A1 (2019).
  • Suven life sciences ltd. Muscarinic M1 receptor positive allosteric modulators. US2019343812A1 (2019).
  • Suven life sciences ltd. Substituted azacycles as muscarinic M1 receptor positive allosteric modulators. WO2019102365A1 (2019).
  • Suven life sciences ltd.Pyrrolo-pyridazine derivatives as muscarinic M1 receptor positive allosteric modulators. WO2020079606A1 (2020).
  • Suven life sciences ltd. Heteroaryl compounds as muscarinic M1 receptor positive allosteric modulators. WO2019077517A1 (2019).
  • Fisher A. Cholinergic modulation of amyloid precursor protein processing with emphasis on M1 muscarinic receptor: perspectives and challenges in treatment of Alzheimer’s disease. J Neurochem. 2012;120:22–33.
  • Melancon BJ, Tarr JC, Panarese JD, et al. Allosteric modulation of the M1 muscarinic acetylcholine receptor: improving cognition and a potential treatment for schizophrenia and Alzheimer’s disease. Drug Discov Today. 2013;18(23–24):1185–1199.
  • Moran SP, Maksymetz J, Conn PJ. Targeting muscarinic acetylcholine receptors for the treatment of psychiatric and neurological disorders. Trends Pharmacol Sci. 2019;40(12):1006–1020. .
  • Felder CC, Goldsmith PJ, Jackson K, et al. Current status of muscarinic M1 and M4 receptors as drug targets for neurodegenerative diseases. Neuropharmacology. 2018;136:449–458.
  • Chase Pharmaceuticals Corp. Oxybutynin transdermal therapeutic system combination. US10149828B2.(2018).
  • McCrery RJ, Appell RA. Oxybutynin: an overview of the available formulations.. Ther Clin Risk Manag. 2006;2(1):19.
  • Sink KM, Thomas III J, Xu H, et al. Dual use of bladder anticholinergics and cholinesterase inhibitors: long-term functional and cognitive outcomes. J Am Geriatr Soc. 2008;56(5):847–853.
  • Suven life sciences ltd. Combination of histamine-3 receptor inverse agonists with acetylcholinesterase inhibitors. WO2018033847A1 (2018).
  • Suven life sciences ltd. Triple combination of histamine-3 receptor inverse agonists, acetylcholinesterase inhibitors and NMDA receptor antagonist. AU2017311666A1 (2019).
  • Vohora D, Bhowmik M. Histamine H3 receptor antagonists/inverse agonists on cognitive and motor processes: relevance to Alzheimer’s disease, ADHD, schizophrenia, and drug abuse. Front Syst Neurosci. 2012;6:72.
  • Witkin J, Nelson D. Selective histamine H3 receptor antagonists for treatment of cognitive deficiencies and other disorders of the central nervous system. Pharmacol Ther. 2004;103(1):1–20.
  • Ghamari N, Zarei O, Arias-Montaño J-A, et al. Histamine H3 receptor antagonists/inverse agonists: where do they go? Pharmacol Ther. 2019;200:69–84.
  • Richter gedeon nyrt. H3 antagonists containing phenoxypiperidine core structure. US9994525B2 (2018).
  • Suven life sciences ltd. Combination of pure 5-HT6 receptor antagonists with acetylcholinesterase inhibitors. CA3023819A1 (2017).
  • Suven life sciences ltd. Triple combination of pure 5-HT6 receptor antagonists, acetylcholinesterase inhibitors and NMDA receptor antagonist. US2019160055A1 (2019).
  • Ferrero H, Solas M, Francis PT, et al. Serotonin 5-HT6 receptor antagonists in alzheimer’s disease: therapeutic rationale and current development status. CNS Drugs. 2017;31(1):19–32.
  • Andrews M, Tousi B, Sabbagh MN. 5HT6 antagonists in the treatment of Alzheimer’s dementia: current progress. Neurol Ther. 2018;7(1):51–58.
  • Suven life sciences ltd. N-arylsulfonyl-3-substituted indoles having serotonin receptor affinity, process for their preparation and pharmaceutical composition containing them. US7875605B2 (2011).
  • Lundbeck H, A S 5-HT6 receptor antagonists for use in the treatment of alzheimer’s disease with apathy as comorbiity. US2017326107A1·(2017).
  • Lundbeck H, A S. Methods of treating Alzheimer’s disease and pharmaceutical compositions thereof. JP. 2019;2019059760A.
  • Lundbeck H, A S Combination of a 5-HT6 receptor antagonist and an acetylcholinesterase inhibitor for use in the treatment of Alzheimer’s disease in a patient subpopulation carrying apoe4 alleles. WO2018215478A1(2018).
  • Axovant sciences gmbH. Methods of treating a neurodegenerative disease. US2019111052A1.(2019).
  • Inc T. 5-HT4 receptor agonist compounds for treatment of cognitive disorders. CN. 2016;105832735A.
  • University of Caen. Combination of acetylcholinesterase inhibitor and 5-HT4 receptor agonist as neuroprotective agent in the treatment of neurodegenerative diseases. WO2020064979A1 (2020).
  • Eglen R, Bonhaus D, Johnson L, et al. Pharmacological characterization of two novel and potent 5-HT4 receptor agonists, RS 67333 and RS 67506, in vitro and in vivo. Br J Pharmacol. 1995;115(8):1387–1392.
  • Fechter Cary Erwin. Combination medication for neuro-degenerative diseases. . CA2990396A1.(2016).
  • Commissariat Energie Atomique. Use of anti-connexin agents for enhancing the therapeutic effect of acetylcholinesterase inhibitors. US2018177773A1 (2018).
  • Harks EG, de Roos AD, Peters PH, et al. Fenamates: a novel class of reversible gap junction blockers. J Pharmacol Exp Ther. 2001;298(3):1033–1041.
  • Juszczak GR, Swiergiel AH. Properties of gap junction blockers and their behavioural, cognitive and electrophysiological effects: animal and human studies. Prog Neuro Psychopharmacol Biol Psychiatry. 2009;33(2):181–198.
  • Yao J, Morioka T, Oite T. PDGF regulates gap junction communication and connexin43 phosphorylation by PI 3-kinase in mesangial cells. Kidney Int. 2000;57(5):1915–1926.
  • Imbimbo BP. An update on the efficacy of non-steroidal anti-inflammatory drugs in Alzheimer’s disease. Expert Opin Investig Drugs. 2009;18(8):1147–1168.
  • Agenebio Inc. Methods and compositions for improving cognitive function. US2016271108A1 (2016).
  • Stout KA, Dunn AR, Hoffman C, et al. The synaptic vesicle glycoprotein 2: structure, function, and disease relevance. ACS Chem Neurosci. 2019;10(9):3927–3938.
  • Steinhoff BJ, Staack AM. Levetiracetam and brivaracetam: a review of evidence from clinical trials and clinical experience. Ther Adv Neurol Disord. 2019;12:1756286419873518.
  • Borghans LGJM, Sambeth A, Prickaerts J, et al. The effects of the soluble guanylate cyclase stimulator riociguat on memory performance in healthy volunteers with a biperiden-induced memory impairment. Psychopharmacology (Berl). 2018;235(8):2407–2416.
  • Acad Ziekenhuis Maastricht. Treatment of cognitive impairment with CGC stimulator. WO2017108441A1 (2017).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.